1
|
Hu S, Shen F, Jia N, Zhang C, Wu X, Jiang W, Li B, Chen Q. Novel monomers recipe derived from Shengji-Huayu formula targeting PI3K/Akt signaling pathway for diabetic wound healing based on accurate network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119509. [PMID: 39971014 DOI: 10.1016/j.jep.2025.119509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/03/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shengji-Huayu (SJHY) formula has been used clinically for diabetic ulcer (DU) treatment. The transcriptional profiles analysis revealed the PI3K/Akt signaling pathway plays a critical role for diabetic wound healing. However, the effects and underlying mechanisms of SJHY are often challenging in diabetic wound treatment. AIM OF THE STUDY To explore the novel monomers recipe and mechanism of SJHY treated diabetic wound via the in vivo and in vitro experiments. MATERIALS AND METHODS The diabetic wound mice model was established to evaluate the therapeutic efficacy of SJHY treated diabetic ulcer. The transcriptional profile was implemented to identify the differentially expressed genes (DEGs) of SJHY treated diabetic wound mice. The constructed PPI network and KEGG-target network were used to identify the core pathway and related targets. The HPLC-MS method was used to identify the ingredients of SJHY formula. The molecular docking and in vitro experiment was used to screen which monomers regulated core pathway in diabetic wound. Finally, a novel monomers recipe was performed for diabetic wound healing. RESULTS The in vivo experiment demonstrated SJHY formula significantly promoted diabetic wound healing. Transcriptomic and network analysis revealed the existence of PI3K/Akt signaling pathway was high correlated with SJHY treated diabetic wound. The HPLC-MS and molecular docking revealed that Calycosin (Cal) and Dehydromiltirone (DHT) were strongly targeting Itga6 and Thbs1. The mRNA and protein levels suggested that Itga6 and Thbs1 acted as important upstream regulators of PI3K/Akt signaling pathway in diabetic wound, and the in vitro experiments revealed Cal, DHT and their recipe were significantly increased the levels of Itga6, Thbs1, PI3K and Akt in MGO induced HaCaT cells. CONCLUSIONS SJHY formula has therapeutic efficiency for diabetic wound healing, and Cal and DHT may be a part of the important monomers that alleviate inflammation via activating the PI3K/Akt signaling pathway by regulated Itga6 and Thbs1. Our study demonstrated Cal and DHT formed a novel monomers recipe, which may be one of the critical strategies of SJHY formula promote diabetic wound healing.
Collapse
Affiliation(s)
- Sheng Hu
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Fang Shen
- Department of Traditional Chinese Medicine Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Ning Jia
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Caiyun Zhang
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Xinxin Wu
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Wencheng Jiang
- Department of Traditional Chinese Medicine Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| | - Bin Li
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| | - Qilong Chen
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| |
Collapse
|
2
|
Liu Z, Bian X, Luo L, Björklund ÅK, Li L, Zhang L, Chen Y, Guo L, Gao J, Cao C, Wang J, He W, Xiao Y, Zhu L, Annusver K, Gopee NH, Basurto-Lozada D, Horsfall D, Bennett CL, Kasper M, Haniffa M, Sommar P, Li D, Landén NX. Spatiotemporal single-cell roadmap of human skin wound healing. Cell Stem Cell 2025; 32:479-498.e8. [PMID: 39729995 DOI: 10.1016/j.stem.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/24/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024]
Abstract
Wound healing is vital for human health, yet the details of cellular dynamics and coordination in human wound repair remain largely unexplored. To address this, we conducted single-cell multi-omics analyses on human skin wound tissues through inflammation, proliferation, and remodeling phases of wound repair from the same individuals, monitoring the cellular and molecular dynamics of human skin wound healing at an unprecedented spatiotemporal resolution. This singular roadmap reveals the cellular architecture of the wound margin and identifies FOSL1 as a critical driver of re-epithelialization. It shows that pro-inflammatory macrophages and fibroblasts sequentially support keratinocyte migration like a relay race across different healing stages. Comparison with single-cell data from venous and diabetic foot ulcers uncovers a link between failed keratinocyte migration and impaired inflammatory response in chronic wounds. Additionally, comparing human and mouse acute wound transcriptomes underscores the indispensable value of this roadmap in bridging basic research with clinical innovations.
Collapse
Affiliation(s)
- Zhuang Liu
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Xiaowei Bian
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Lihua Luo
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Åsa K Björklund
- Department of Life Science, National Bioinformatics Infrastructure Sweden, Göteborg, Sweden; Science for Life Laboratory, Chalmers University of Technology, 41296 Göteborg, Sweden
| | - Li Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Letian Zhang
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Lei Guo
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Juan Gao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Chunyan Cao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Jiating Wang
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Wenjun He
- The first affiliated hospital of Soochow University, Department of Plastic and Burn Surgery. NO.188, Shizi Street, Suzhou, Jiangsu, China
| | - Yunting Xiao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Liping Zhu
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Karl Annusver
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Daniela Basurto-Lozada
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - David Horsfall
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Clare L Bennett
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Pehr Sommar
- Department of Plastic and Reconstructive Surgery, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Dongqing Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China.
| | - Ning Xu Landén
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden.
| |
Collapse
|
3
|
Huang J, Deng Q, Tsang LL, Chang G, Guo J, Ruan YC, Wang CC, Li G, Chan HF, Zhang X, Jiang X. Mesenchymal stem cells from perinatal tissues promote diabetic wound healing via PI3K/AKT activation. Stem Cell Res Ther 2025; 16:59. [PMID: 39923118 PMCID: PMC11807333 DOI: 10.1186/s13287-025-04141-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/13/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Diabetic foot ulcers (DFUs) represent a major complication of diabetes, often leading to poor healing outcomes with conventional treatments. Mesenchymal stem cell (MSC) therapies have emerged as a promising alternative, given their potential to modulate various pathways involved in wound healing. This study evaluates and compares the therapeutic potential of MSCs derived from perinatal tissues-human umbilical cord MSCs (hUCMSCs), human chorionic villi MSCs (hCVMSCs), and human decidua basalis MSCs (hDCMSCs)-in a diabetic wound healing model. METHODS We performed in vitro and in vivo studies to compare the efficacy of hUCMSCs, hCVMSCs, and hDCMSCs. Mass spectrometry was used to analyze the secreted proteins of the MSCs. We incorporated the MSCs into a polyethylene glycol diacrylate (PEGDA) and sodium alginate (SA) hydrogel matrix with collagen I (Col-I) to evaluate their effects on wound healing. RESULTS All three types of MSCs promoted wound healing, with hUCMSCs and hCVMSCs showing stronger effects compared to hDCMSCs. Both hUCMSCs and hCVMSCs demonstrated robust wound healing kinetics, with enhanced keratinocyte proliferation (KRT14+/Ki67+ cells), maturation (KRT10/KRT14 ratio), and angiogenesis. In vitro studies demonstrated that the MSC-derived secretome enhanced keratinocyte proliferation and migration, endothelial cell function and stem cell recruitment, indicating robust paracrine effects. Mass spectrometry revealed a conserved set of proteins including THBS1 (thrombospondin 1), SERPINE1 (serpin family E member 1), ANXA1 (annexin A1), LOX (lysyl oxidase), and ITGB1 (integrin beta-1) which are involved in extracellular matrix (ECM) organization and wound healing, with the PI3K/AKT signaling pathway playing a central role. The PEGDA/SA/Col-I hydrogel demonstrated a unique balance of mechanical and biological properties and an optimal environment for MSC viability and function. Application of either hUCMSC- or hCVMSC-laden hydrogels resulted in accelerated wound closure, improved re-epithelialization, increased collagen deposition, and enhanced vascularization in vivo. CONCLUSIONS MSCs From perinatal tissues particularly hUCMSCs and hCVMSCs significantly enhance diabetic wound healing through PI3K/AKT pathway activation while hDCMSCs exhibited weaker efficacy. The PEGDA/SA/Col-I hydrogel supports MSC viability and function offering a promising scaffold for DFU treatment. These findings underscore the potential of specific perinatal MSCs and optimized hydrogel formulations in advancing diabetic wound care.
Collapse
Affiliation(s)
- Jiawei Huang
- School of Biomedical Sciences, Faculty of Medicine; CUHK-GIBH CAS Joint Research Laboratory On Stem Cell and Regenerative Medicine; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qingwen Deng
- School of Biomedical Sciences, Faculty of Medicine; CUHK-GIBH CAS Joint Research Laboratory On Stem Cell and Regenerative Medicine; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lai Ling Tsang
- School of Biomedical Sciences, Faculty of Medicine; CUHK-GIBH CAS Joint Research Laboratory On Stem Cell and Regenerative Medicine; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Guozhu Chang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jinghui Guo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, China
| | - Ye Chun Ruan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong; Reproduction and Development, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiaohu Zhang
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Xiaohua Jiang
- School of Biomedical Sciences, Faculty of Medicine; CUHK-GIBH CAS Joint Research Laboratory On Stem Cell and Regenerative Medicine; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, The Chinese University of Hong Kong, Hong Kong SAR, China.
- The Chinese University of Hong Kong, Shenzhen Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
4
|
Wang Y, Hess ME, Tan Y, Esser PR, Nyström A, Boerries M, Sayar SB, Has C. Alterations in the microenvironment of junctional epidermolysis bullosa keratinocytes: A gene expression study. Matrix Biol 2025; 135:12-23. [PMID: 39615637 DOI: 10.1016/j.matbio.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/12/2024] [Accepted: 11/24/2024] [Indexed: 01/15/2025]
Abstract
Integrin α6β4 subunits and type XVII collagen are critical transmembrane proteins involved in cell-matrix adhesion in skin, while laminin 332 serves as their ligand in the basement membrane zone (BMZ). Those proteins contribute to the composition of hemidesmosomes (HDs) and pathogenic variants in their corresponding genes cause junctional epidermolysis bullosa (JEB). Although the genotype-phenotype relationships in JEB have been extensively studied, the pathogenetic changes of extracellular matrix (ECM) and cell-matrix adhesion resulting from gene mutations remain unclear. We conducted a global unbiased transcriptome analysis using bulk RNA sequencing (RNA-seq) on selected JEB donor-derived cell lines lacking integrin β4 subunit (ITGB4-), type XVII collagen (COL17-) and laminin β3 chain (LAMB3-), respectively. Additional JEB cell lines and JEB donor skin samples were used for validation of relevant findings. Collectively, the results revealed similar dysregulation patterns of ECM and focal adhesion (FAs) associated genes in ITGB4- and COL17- cell lines, while LAMB3- cells displayed a relatively opposite tendency. Importantly, key nodes in the dysregulated network were associated with ECM proteins involved in wound healing processes. Additionally, a group of inflammatory-associated genes was disclosed to be up-regulated in JEB keratinocytes and could not be normalized by the adhesion rescue. The functional assay further revealed the hierarchy of stable adhesion among mutant cell lines COL17->ITGB4->LAMB3-, which correlates with the severity of their clinical manifestations. Our results indicated a wound healing associated ECM and inflammatory microenvironment established by JEB keratinocytes.
Collapse
Affiliation(s)
- Yao Wang
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Maria Elena Hess
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Yan Tan
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Philipp R Esser
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Comprehensive Cancer Centre Freiburg (CCCF), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; German Cancer Consortium (DKTK), a partnership between DKFZ and Medical Center - University of Freiburg, Freiburg, Germany
| | - Saliha Beyza Sayar
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Cristina Has
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
5
|
Wang L, Guo W, Tian Y, Wang J, Xu S, Shu W, Liang H, Chen M. Carboxypeptidase inhibitor Latexin (LXN) regulates intestinal organogenesis and intestinal remodeling involved in intestinal injury repair in mice. Int J Biol Macromol 2024; 279:135129. [PMID: 39208900 DOI: 10.1016/j.ijbiomac.2024.135129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/10/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The self-renewal and regeneration of intestinal epithelium are mainly driven by intestinal stem cells resided in crypts, which are crucial for rapid recovery intestinal tissue following injury. Latexin (LXN) is a highly expressed stem cell proliferation and differentiation related gene in intestinal tissue. However, it is still ambiguous whether LXN participates in intestine regeneration by regulating intestinal stem cells (ISCs). Here, we report that LXN colocalizes with Leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) in intestinal crypts, and deletion of LXN upregulates the expression of Lgr5 in intestinal crypts. LXN deficiency promotes the proliferation of ISCs, thereby enhances the development of intestinal organoids. Mechanically, we show that LXN deficiency enhances the expression of Lgr5 in ISCs by activating the Yes-associated protein (YAP) and wingless (Wnt) signal pathways, thus accelerating intestinal normal growth and regeneration post-injury. In summary, these findings uncover a novel function of LXN in intestinal regeneration post-injury and intestinal organogenesis, suggesting the potential role of LXN in the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Lingzhu Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Wenwen Guo
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Yang Tian
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Jingzhu Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Shaohua Xu
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Wei Shu
- College of Biotechnology, Guilin Medical University, Guilin, China.
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.
| | - Ming Chen
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.
| |
Collapse
|
6
|
Wang M, Hong Y, Fu X, Sun X. Advances and applications of biomimetic biomaterials for endogenous skin regeneration. Bioact Mater 2024; 39:492-520. [PMID: 38883311 PMCID: PMC11179177 DOI: 10.1016/j.bioactmat.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 06/18/2024] Open
Abstract
Endogenous regeneration is becoming an increasingly important strategy for wound healing as it facilitates skin's own regenerative potential for self-healing, thereby avoiding the risks of immune rejection and exogenous infection. However, currently applied biomaterials for inducing endogenous skin regeneration are simplistic in their structure and function, lacking the ability to accurately mimic the intricate tissue structure and regulate the disordered microenvironment. Novel biomimetic biomaterials with precise structure, chemical composition, and biophysical properties offer a promising avenue for achieving perfect endogenous skin regeneration. Here, we outline the recent advances in biomimetic materials induced endogenous skin regeneration from the aspects of structural and functional mimicry, physiological process regulation, and biophysical property design. Furthermore, novel techniques including in situ reprograming, flexible electronic skin, artificial intelligence, single-cell sequencing, and spatial transcriptomics, which have potential to contribute to the development of biomimetic biomaterials are highlighted. Finally, the prospects and challenges of further research and application of biomimetic biomaterials are discussed. This review provides reference to address the clinical problems of rapid and high-quality skin regeneration.
Collapse
Affiliation(s)
- Mengyang Wang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
| | - Yiyue Hong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| |
Collapse
|
7
|
Wang M, Zan T, Fan C, Li Z, Wang D, Li Q, Zhang C. Advances in GPCR-targeted drug development in dermatology. Trends Pharmacol Sci 2024; 45:678-690. [PMID: 39060127 DOI: 10.1016/j.tips.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Achieving the efficacy and specificity of G-protein-coupled receptor (GPCR) targeting-drugs in the skin remains challenging. Understanding the molecular mechanism underlying GPCR dysfunction is crucial for developing targeted therapies. Recent advances in genetic, signal transduction, and structural studies have significantly improved our understanding of cutaneous GPCR functions in both normal and pathological states. In this review, we summarize recent discoveries of pathogenic GPCRs in dermal injuries, chronic inflammatory dermatoses, cutaneous malignancies, as well as the development of potent potential drugs. We also discuss targeting of cutaneous GPCR complexes via the transient receptor potential (TRP) channel and structure elucidation, which provide new opportunities for therapeutic targeting of GPCRs involved in skin disorders. These insights are expected to lead to more effective and specific treatments for various skin conditions.
Collapse
Affiliation(s)
- Meng Wang
- Songjiang Research Institute, Songjiang Hospital, affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Chengang Fan
- Department of Orthopedics and Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhouxiao Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Danru Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Chao Zhang
- Department of Orthopedics and Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
8
|
Li WZ, Liu XX, Shi YJ, Wang XR, Li L, Tai ML, Yi F. Unveiling the mechanism of high sugar diet induced advanced glycosylation end products damage skin structure via extracellular matrix-receptor interaction pathway. J Cosmet Dermatol 2024; 23:2496-2508. [PMID: 38501159 DOI: 10.1111/jocd.16295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/09/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND AGEs accumulate in the skin as a result of a high-sugar diet and play an important role in the skin aging process. OBJECTIVES The aim of this study was to characterize the mechanism underlying the effect of a high-sugar diet on skin aging damage at a holistic level. METHODS We established a high-sugar diet mouse model to compare and analyze differences in physiological indexes. The effect of a high-sugar diet on skin aging damage was analyzed by means of a transcriptome study and staining of pathological sections. Furthermore, the differences in the protein expression of AGEs and ECM components between the HSD and control groups were further verified by immunohistochemistry. RESULTS The skin in the HSD group mice tended toward a red, yellow, dark, and deep color. In addition, the epidermis was irregular with anomalous phenomena, the epidermis was thinned, and the dermis lost its normal structure and showed vacuolated changes. Transcriptomics results revealed significant downregulation of the ECM-receptor interaction pathway, significant upregulation of the expression of AGEs and significant downregulation of the expression levels of COLI, FN1, LM5, and TNC, among others ECM proteins and ECM receptors. CONCLUSIONS High-sugar diets cause skin aging damage by inducing the accumulation of AGEs, disrupting the expression of ECM proteins and their receptors, and downregulating the ECM-receptor interaction pathway, which affects cellular behavioral functions such as cell proliferation, migration, and adhesion, as well as normal skin tissue structure.
Collapse
Affiliation(s)
- Wan-Zhao Li
- R&D Center, Infinitus (China) Company Ltd, Guangzhou, China
| | - Xiao-Xing Liu
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China
| | - Yu-Jing Shi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao-Rui Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China
| | - Mei-Ling Tai
- R&D Center, Infinitus (China) Company Ltd, Guangzhou, China
| | - Fan Yi
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
9
|
Raja E, Clarin MTRDC, Yanagisawa H. Matricellular Proteins in the Homeostasis, Regeneration, and Aging of Skin. Int J Mol Sci 2023; 24:14274. [PMID: 37762584 PMCID: PMC10531864 DOI: 10.3390/ijms241814274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Matricellular proteins are secreted extracellular proteins that bear no primary structural functions but play crucial roles in tissue remodeling during development, homeostasis, and aging. Despite their low expression after birth, matricellular proteins within skin compartments support the structural function of many extracellular matrix proteins, such as collagens. In this review, we summarize the function of matricellular proteins in skin stem cell niches that influence stem cells' fate and self-renewal ability. In the epidermal stem cell niche, fibulin 7 promotes epidermal stem cells' heterogeneity and fitness into old age, and the transforming growth factor-β-induced protein ig-h3 (TGFBI)-enhances epidermal stem cell growth and wound healing. In the hair follicle stem cell niche, matricellular proteins such as periostin, tenascin C, SPARC, fibulin 1, CCN2, and R-Spondin 2 and 3 modulate stem cell activity during the hair cycle and may stabilize arrector pili muscle attachment to the hair follicle during piloerections (goosebumps). In skin wound healing, matricellular proteins are upregulated, and their functions have been examined in various gain-and-loss-of-function studies. However, much remains unknown concerning whether these proteins modulate skin stem cell behavior, plasticity, or cell-cell communications during wound healing and aging, leaving a new avenue for future studies.
Collapse
Affiliation(s)
- Erna Raja
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
| | - Maria Thea Rane Dela Cruz Clarin
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
- Ph.D. Program in Humanics, School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
| |
Collapse
|
10
|
Li Y, Hashikawa K, Ebisawa K, Kambe M, Higuchi S, Kamei Y. <Editors' Choice> Supernatant from activated omentum accelerates wound healing in diabetic mice wound model. NAGOYA JOURNAL OF MEDICAL SCIENCE 2023; 85:528-541. [PMID: 37829482 PMCID: PMC10565575 DOI: 10.18999/nagjms.85.3.528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/04/2022] [Indexed: 10/14/2023]
Abstract
Diabetic wounds are considered one of the most frequent and severe complications of diabetes mellitus. Recently, the omentum has been used in diabetic wound healing because of its tissue repair properties. The activated omentum is richer in growth factors than the inactivated, thereby contributing to the wound healing process. To further investigate the effect of activated omentum conditioned medium (aOCM) on diabetic wound healing, we injected supernatant from aOCM, saline-OCM (sOCM), inactivated-OCM (iOCM), and medium (M) subcutaneously upon creation of a cutaneous wound healing model in diabetic mice. Wound area (%) was evaluated on days 0, 3, 5, 7, 9, 11, 14, 21, and 28 post-operation. At 9 and 28 d post-operation, skin tissue was harvested and assessed for gross observation, neovascularization, peripheral nerve fiber regeneration, and collagen deposition. We observed that aOCM enhanced the wound repair process, with significant acceleration of epidermal and collagen deposition in the surgical lesion on day 9. Additionally, aOCM displayed marked efficiency in neovascularization and peripheral nerve regeneration during wound healing. Thus, aOCM administration exerts a positive influence on the diabetic mouse model, which can be employed as a new therapy for diabetic wounds.
Collapse
Affiliation(s)
- Yu Li
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazunobu Hashikawa
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsumi Ebisawa
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Miki Kambe
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Higuchi
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuzuru Kamei
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
11
|
Tearle JL, Arjunan SN, Tay SS, Colakoglu F, Cremasco J, Golo M, Biro M. Targeted Single-cell Isolation of Spontaneously Escaping Live Melanoma Cells for Comparative Transcriptomics. CANCER RESEARCH COMMUNICATIONS 2023; 3:1524-1537. [PMID: 37575281 PMCID: PMC10416804 DOI: 10.1158/2767-9764.crc-22-0305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 05/30/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023]
Abstract
Solid cancer cells escape the primary tumor mass by transitioning from an epithelial-like state to an invasive migratory state. As they escape, metastatic cancer cells employ interchangeable modes of invasion, transitioning between fibroblast-like mesenchymal movement to amoeboid migration, where cells display a rounded morphology and navigate the extracellular matrix in a protease-independent manner. However, the gene transcripts that orchestrate the switch between epithelial, mesenchymal, and amoeboid states remain incompletely mapped, mainly due to a lack of methodologies that allow the direct comparison of the transcriptomes of spontaneously invasive cancer cells in distinct migratory states. Here, we report a novel single-cell isolation technique that provides detailed three-dimensional data on melanoma growth and invasion, and enables the isolation of live, spontaneously invasive cancer cells with distinct morphologies and invasion parameters. Via the expression of a photoconvertible fluorescent protein, compact epithelial-like cells at the periphery of a melanoma mass, elongated cells in the process of leaving the mass, and rounded amoeboid cells invading away from the mass were tagged, isolated, and subjected to single-cell RNA sequencing. A total of 462 differentially expressed genes were identified, from which two candidate proteins were selected for further pharmacologic perturbation, yielding striking effects on tumor escape and invasion, in line with the predictions from the transcriptomics data. This work describes a novel, adaptable, and readily implementable method for the analysis of the earliest phases of tumor escape and metastasis, and its application to the identification of genes underpinning the invasiveness of malignant melanoma. Significance This work describes a readily implementable method that allows for the isolation of individual live tumor cells of interest for downstream analyses, and provides the single-cell transcriptomes of melanoma cells at distinct invasive states, both of which open avenues for in-depth investigations into the transcriptional regulation of the earliest phases of metastasis.
Collapse
Affiliation(s)
- Jacqueline L.E. Tearle
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
- Present address: Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Satya N.V. Arjunan
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
- Present address: Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Szun S. Tay
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Feyza Colakoglu
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - James Cremasco
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
- Present address: Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Matteo Golo
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Maté Biro
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
12
|
Ievlev V, Pai AC, Dillon DS, Kuhl S, Lynch TJ, Freischlag KW, Gries CB, Engelhardt JF, Parekh KR. Development and characterization of ferret ex vivo tracheal injury and cell engraftment model. Front Med (Lausanne) 2023; 10:1144754. [PMID: 37113613 PMCID: PMC10126424 DOI: 10.3389/fmed.2023.1144754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/15/2023] [Indexed: 04/29/2023] Open
Abstract
The field of airway biology research relies primarily on in vitro and in vivo models of disease and injury. The use of ex vivo models to study airway injury and cell-based therapies remains largely unexplored although such models have the potential to overcome certain limitations of working with live animals and may more closely replicate in vivo processes than in vitro models can. Here, we characterized a ferret ex vivo tracheal injury and cell engraftment model. We describe a protocol for whole-mount staining of cleared tracheal explants, and showed that it provides a more comprehensive structural overview of the surface airway epithelium (SAE) and submucosal glands (SMGs) than 2D sections, revealing previously underappreciated structural anatomy of tracheal innervation and vascularization. Using an ex vivo model of tracheal injury, we evaluated the injury responses in the SAE and SMGs that turned out to be consistent with published in vivo work. We used this model to assess factors that influence engraftment of transgenic cells, providing a system for optimizing cell-based therapies. Finally, we developed a novel 3D-printed reusable culture chamber that enables live imaging of tracheal explants and differentiation of engrafted cells at an air-liquid interface. These approaches promise to be useful for modeling pulmonary diseases and testing therapies. Graphical abstract1,2. We describe here a method for differential mechanical injury of ferret tracheal explants that can be used to evaluate airway injury responses ex vivo. 3. Injured explants can be cultured at ALI (using the novel tissue-transwell device on the right) and submerged long-term to evaluate tissue-autonomous regeneration responses. 4. Tracheal explants can also be used for low throughput screens of compounds to improve cell engraftment efficiency or can be seeded with particular cells to model a disease phenotype. 5. Lastly, we demonstrate that ex vivo-cultured tracheal explants can be evaluated by various molecular assays and by immunofluorescent imaging that can be performed live using our custom-designed tissue-transwell.
Collapse
Affiliation(s)
- Vitaly Ievlev
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Albert C. Pai
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Drew S. Dillon
- Protostudios, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Spencer Kuhl
- Protostudios, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Thomas J. Lynch
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Kyle W. Freischlag
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Caitlyn B. Gries
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Kalpaj R. Parekh
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| |
Collapse
|
13
|
Zhang JT, Wu MF, Ma MH, Zhao L, Zhu JY, Nian H, Li FL. Research on the wound healing effect of Shengji Huayu Formula ethanol extract-derived fractions in streptozotocin-induced diabetic ulcer rats. BMC Complement Med Ther 2023; 23:67. [PMID: 36859252 PMCID: PMC9976525 DOI: 10.1186/s12906-023-03894-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 02/17/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Diabetic ulcer is a common complication of diabetes. It is characterized by a long-term disease course and high recurrence rate. Shengji Huayu Formula (SHF) is an effective formula for treating diabetic ulcers. However, the specific effective parts of SHF remain unclear. Clarifying the active polar site of SHF would be helpful to refine research on the components in SHF that promote wound healing. This research aims to focus on evaluating the activity of polar fractions. METHODS A diabetic rat model was established by intraperitoneally injecting streptozotocin (STZ) and was adopted to confirm the therapeutic effect of SHF. Four different polarity parts were extracted from SHF and prepared into a cream to evaluate the activity. High-performance liquid chromatography (HPLC) was used to detect chemical constituents in chloroform extracts. RESULTS It was discovered that dracorhodin, aloe-emodin, rhein, imperatorin, emodin, isoimperatorin, chrysophanol, physcion, and tanshinone IIA were the main components of the chloroform extract from SHF. The results revealed that chloroform extract could effectively accelerate diabetic wound healing by promoting collagen regeneration and epidermal repair. Chloroform extract of SHF could stimulate the generation of vascular endothelial growth factor (VEGF). The results are also indicated that the effective active fraction was the chloroform part, and the method of detecting the main chemical constituents in the active part was successfully established. CONCLUSION SHF could improve diabetic ulcers by promoting granulation tissue synthesis. In this study, four polar parts (petroleum ether, chloroform, ethylacetate, n-butanol) were extracted from a 95% ethanol extract. In contrast, chloroform polar parts showed a higher wound closure rate, stimulated more collagen regeneration and promoted more production of vascular endothelial cells. In conclusion, the chloroform extract of SHF was the effective polar part in ameliorating diabetic wound healing.
Collapse
Affiliation(s)
- Jing-Ting Zhang
- grid.412540.60000 0001 2372 7462Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437 China ,grid.412540.60000 0001 2372 7462Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Yueyang, 200437 China ,grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University, Shanghai, 200080 China
| | - Min-Feng Wu
- grid.8547.e0000 0001 0125 2443Department of Dermatology, Huadong Hospital, Fudan University, Shanghai, 200040 China
| | - Ming-Hua Ma
- grid.460149.e0000 0004 1798 6718Department of Pharmacy, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090 China
| | - Liang Zhao
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, 201908 China
| | - Jian-Yong Zhu
- grid.412540.60000 0001 2372 7462Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437 China
| | - Hua Nian
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Yueyang, 200437, China.
| | - Fu-Lun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| |
Collapse
|
14
|
Peres C, Sellitto C, Nardin C, Putti S, Orsini T, Di Pietro C, Marazziti D, Vitiello A, Calistri A, Rigamonti M, Scavizzi F, Raspa M, Zonta F, Yang G, White TW, Mammano F. Antibody gene transfer treatment drastically improves epidermal pathology in a keratitis ichthyosis deafness syndrome model using male mice. EBioMedicine 2023; 89:104453. [PMID: 36736132 PMCID: PMC9926223 DOI: 10.1016/j.ebiom.2023.104453] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/30/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Keratitis ichthyosis deafness (KID) syndrome is a rare disorder caused by hemichannel (HC) activating gain-of-function mutations in the GJB2 gene encoding connexin (Cx) 26, for which there is no cure, or current treatments based upon the mechanism of disease causation. METHODS We applied Adeno Associated Virus (AAV) mediated mAb gene transfer (AAVmAb) to treat the epidermal features of KID syndrome with a well-characterized HC blocking antibody using male mice of a murine model that replicates the skin pathology of the human disease. FINDINGS We demonstrate that in vivo AAVmAb treatment significantly reduced the size and thickness of KID lesions, in addition to blocking activity of mutant HCs in the epidermis in vivo. We also show that AAVmAb treatment eliminated abnormal keratinocyte proliferation and enlarged cell size, decreased apoptosis, and restored the normal distribution of keratin expression. INTERPRETATION Our findings reinforce the critical role played by increased HC activity in the skin pathology associated with KID syndrome. They also underscore the clinical potential of anti-HC mAbs coupled with genetic based delivery systems for treating the underlying mechanistic basis of this disorder. Inhibition of HC activity is an ideal therapeutic target in KID syndrome, and the genetic delivery of mAbs targeted against mutant HCs could form the basis of new therapeutic interventions to treat this incurable disease. FUNDING Fondazione Telethon grant GGP19148 and University of Padova grant Prot. BIRD187130 to FM; Foundation for Ichthyosis and Related Skin Types (FIRST) and National Institutes of Health grant EY 026911 to TWW.
Collapse
Affiliation(s)
- Chiara Peres
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy
| | - Caterina Sellitto
- Department of Physiology and Biophysics, Stony Brook University, T5-147, Basic Science Tower; Stony Brook, NY, 11794-8661, USA
| | - Chiara Nardin
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy
| | - Sabrina Putti
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy
| | - Tiziana Orsini
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy
| | - Daniela Marazziti
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy
| | - Adriana Vitiello
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | | | - Ferdinando Scavizzi
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy
| | - Marcello Raspa
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Thomas W White
- Department of Physiology and Biophysics, Stony Brook University, T5-147, Basic Science Tower; Stony Brook, NY, 11794-8661, USA.
| | - Fabio Mammano
- Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Rome, Italy; Department of Physics and Astronomy "G. Galilei", University of Padova, 35131, Padova, Italy.
| |
Collapse
|
15
|
Siriwach R, Ngo AQ, Narumiya S, Thumkeo D. An optimized protocol to identify keratinocyte subpopulations in vitro by single-cell RNA sequencing analysis. STAR Protoc 2022; 3:101906. [PMID: 36595953 PMCID: PMC9730219 DOI: 10.1016/j.xpro.2022.101906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/02/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Here, we describe a protocol for single-cell isolation from the primary culture of normal human epidermal keratinocytes derived from neonatal foreskin. The cell culture conditions have been optimized for inducing expression of keratinocyte differentiation markers. Cells are cultured in the absence or presence of a bioactive lipid lysophosphatidic acid (LPA). Single cells are isolated by Fluidigm C1 system. This is followed by cDNA library preparation using Takara SMART-Seq v4 Ultra and Illumina Nextera XT kit for RNA sequencing. For complete details on the use and execution of this protocol, please refer to Siriwach et al. (2022).1.
Collapse
Affiliation(s)
- Ratklao Siriwach
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Anh Quynh Ngo
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Dean Thumkeo
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan,Corresponding author
| |
Collapse
|
16
|
Cárdenas-León CG, Mäemets-Allas K, Klaas M, Lagus H, Kankuri E, Jaks V. Matricellular proteins in cutaneous wound healing. Front Cell Dev Biol 2022; 10:1073320. [PMID: 36506087 PMCID: PMC9730256 DOI: 10.3389/fcell.2022.1073320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Cutaneous wound healing is a complex process that encompasses alterations in all aspects of the skin including the extracellular matrix (ECM). ECM consist of large structural proteins such as collagens and elastin as well as smaller proteins with mainly regulative properties called matricellular proteins. Matricellular proteins bind to structural proteins and their functions include but are not limited to interaction with cell surface receptors, cytokines, or protease and evoking a cellular response. The signaling initiated by matricellular proteins modulates differentiation and proliferation of cells having an impact on the tissue regeneration. In this review we give an overview of the matricellular proteins that have been found to be involved in cutaneous wound healing and summarize the information known to date about their functions in this process.
Collapse
Affiliation(s)
| | - Kristina Mäemets-Allas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Mariliis Klaas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Heli Lagus
- Department of Plastic Surgery and Wound Healing Centre, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Viljar Jaks
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia,Dermatology Clinic, Tartu University Clinics, Tartu, Estonia,*Correspondence: Viljar Jaks,
| |
Collapse
|